1
|
Zheng J, Zhang J, Han J, Zhao Z, Lin K. The effect of salidroside in promoting endogenous neural regeneration after cerebral ischemia/reperfusion involves notch signaling pathway and neurotrophic factors. BMC Complement Med Ther 2024; 24:293. [PMID: 39090706 PMCID: PMC11295647 DOI: 10.1186/s12906-024-04597-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 07/19/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND Salidroside is the major bioactive and pharmacological active substance in Rhodiola rosea L. It has been reported to have neuroprotective effects on cerebral ischemia/reperfusion (I/R). However, whether salidroside can enhance neural regeneration after cerebral I/R is still unknown. This study investigated the effects of salidroside on the endogenous neural regeneration after cerebral I/R and the related mechanism. METHODS Focal cerebral I/R was induced in rats by transient middle cerebral artery occlusion/reperfusion (MCAO/R). The rats were intraperitoneally treated salidroside once daily for 7 consecutive days. Neurobehavioral assessments were performed at 3 days and 7 days after the injury. TTC staining was performed to assess cerebral infarct volume. To evaluate the survival of neurons, immunohistochemical staining of Neuronal Nuclei (NeuN) in the ischemic hemisphere were conducted. Also, immunofluorescence double or triple staining of the biomarkers of proliferating neural progenitor cells in Subventricular Zone (SVZ) and striatum of the ischemia hemisphere were performed to investigate the neurogenesis. Furthermore, reverse transcription-polymerase chain reaction (RT-PCR) and enzyme-linked immunosorbent assay (ELISA) were used to detect the expression of neurotrophic factors (NTFs) brain-derived neurotrophic factor (BDNF) and nerve growth factor (NGF). Expression of Notch1 and its target molecular Hes1 were also analyzed by western-blotting and RT-PCR. RESULTS Salidroside treatment ameliorated I/R induced neurobehavioral impairment, and reduced infarct volume. Salidroside also restored NeuN positive cells loss after I/R injury. Cerebral I/R injury significantly increased the expression of 5-Bromo-2'-Deoxyuridine (BrdU) and doublecotin (DCX), elevated the number of BrdU/Nestin/DCX triple-labeled cells in SVZ, and BrdU/Nestin/glial fibrillary acidic protein (GFAP) triple-labeled cells in striatum. Salidroside treatment further promoted the proliferation of BrdU/DCX labeled neuroblasts and BrdU/Nestin/GFAP labeled reactive astrocytes. Furthermore, salidroside elevated the mRNA expression and protein concentration of BDNF and NGF in ischemia periphery area, as well. Mechanistically, salidroside elevated Notch1/Hes1 mRNA expression in SVZ. The protein levels of them were also increased after salidroside administration. CONCLUSIONS Salidroside enhances the endogenous neural regeneration after cerebral I/R. The mechanism of the effect may involve the regulation of BDNF/NGF and Notch signaling pathway.
Collapse
Affiliation(s)
- Jiabing Zheng
- Fujian Medical Universtity Union Hospital, Fuzhou, Fujian Province, People's Republic of China
| | - Jizhou Zhang
- Institute of Materia Medica, Fujian Academy of Chinese Medical Sciences, Fuzhou, Fujian Province, People's Republic of China
| | - Jing Han
- Institute of Materia Medica, Fujian Academy of Chinese Medical Sciences, Fuzhou, Fujian Province, People's Republic of China
| | - Zhichang Zhao
- Fujian Medical Universtity Union Hospital, Fuzhou, Fujian Province, People's Republic of China
| | - Kan Lin
- Fujian Medical Universtity Union Hospital, Fuzhou, Fujian Province, People's Republic of China.
| |
Collapse
|
2
|
Ren J, Zhang X, Zhou L, Cao W, Zhang L, Chen X, Li G. Comprehensive evaluation of Dragon's Blood in combination with borneol in ameliorating ischemic/reperfusion brain injury using RNA sequencing, metabolomics, and 16S rRNA sequencing. Front Pharmacol 2024; 15:1372449. [PMID: 38783945 PMCID: PMC11112420 DOI: 10.3389/fphar.2024.1372449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 04/16/2024] [Indexed: 05/25/2024] Open
Abstract
Ischemia/reperfusion (IR) can induce deleterious responses such as apoptosis, inflammation, and oxidative stress; however, there are currently no efficient therapeutics to treat IR brain injury. Dragon's blood (DB) plays a significant role in treating ischemic stroke in China. Borneol (B) is an upper ushering drug that guides drugs to the target organs, including the brain. Therefore, we hypothesized that the combination of DB and B (DB + B) would provide cooperative therapeutic benefits for IR brain injury. To confirm this, we first investigated the protective effect of DB + B in an IR brain injury rat model using the modified neurological severity score (mNSS), infarction size measure, HE staining, and laser speckle contrast imaging. Then, we comprehensively evaluated the mechanism of DB + B in ameliorating IR brain injury based on RNA sequencing, serum untargeted metabolomics, and 16S rRNA sequencing. We have confirmed that DB + B enhanced the efficacy of the ischemic stroke treatment compared to DB or B alone for the first time. Our study provisionally confirms that the mechanism by which DB + B prevents IR brain injury is related to the maintenance of intestinal microecological balance and regulation of metabolic dysfunction, thereby suppressing inflammation and regulating immunity. DB + B may effectively regulate intestinal flora including o_Pseudomonadales, s_Bacteroides_caecimuris, o_unidentified_Bacilli, f-Pseudomonadaceae, and g-Pseudomonas, mainly regulate serum metabolites including improve the protective benefit of IR brain injury lysoPCs and lysoPEs, thus inhibiting TLR4/MyD88/NF-κB and IL-17 signing pathway to reduce inflammatory reactions. hat this mechanism is associated with the maintenance of intestinal flora balance and the regulation of metabolic dysfunction, thereby suppressing inflammation and regulating immunity. This provides scientific support for the clinical translation of DB + B in the prevention and treatment of ischemic stroke and establishes a basis for further investigation of its therapeutic mechanism.
Collapse
Affiliation(s)
- Jiahui Ren
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Yunnan Branch, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Jinghong, China
- Yunnan Key Laboratory of Southern Medicine Utilization, Jinghong, China
| | - Xue Zhang
- Yunnan Key Laboratory of Southern Medicine Utilization, Jinghong, China
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Lingjuan Zhou
- Xishuangbanna Dai Autonomous Prefecture People’s Hospital, Jinghong, China
| | - Wanyu Cao
- Yunnan Key Laboratory of Southern Medicine Utilization, Jinghong, China
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Lixia Zhang
- Yunnan Branch, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Jinghong, China
- Yunnan Key Laboratory of Southern Medicine Utilization, Jinghong, China
| | - Xi Chen
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Yunnan Branch, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Jinghong, China
- Yunnan Key Laboratory of Southern Medicine Utilization, Jinghong, China
| | - Guang Li
- Yunnan Branch, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Jinghong, China
- Yunnan Key Laboratory of Southern Medicine Utilization, Jinghong, China
| |
Collapse
|
3
|
Lim C, Lim S, Moon SJ, Cho S. Neuroprotective effects of methanolic extract from Chuanxiong Rhizoma in mice with middle cerebral artery occlusion-induced ischemic stroke: suppression of astrocyte- and microglia-related inflammatory response. BMC Complement Med Ther 2024; 24:140. [PMID: 38575941 PMCID: PMC10993527 DOI: 10.1186/s12906-024-04454-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 03/25/2024] [Indexed: 04/06/2024] Open
Abstract
BACKGROUND In traditional Asian medicine, dried rhizomes of Ligusticum chuanxiong Hort. (Chuanxiong Rhizoma [CR]) have long been used to treat pain disorders that affect the head and face such as headaches. Furthermore, they have been used primarily for blood circulation improvement or as an analgesic and anti-inflammatory medicine. This study aimed to investigate the neuroprotective effects of a methanol extract of CR (CRex) on ischemic stroke in mice caused by middle cerebral artery occlusion (MCAO). METHODS C57BL/6 mice were given a 1.5-h transient MCAO (MCAO control and CRex groups); CRex was administered in the mice of the CRex group at 1,000-3,000 mg/kg either once (single dose) or twice (twice dose) before MCAO. The mechanism behind the neuroprotective effects of CRex was examined using the following techniques: brain infarction volume, edema, neurological deficit, novel object recognition test (NORT), forepaw grip strength, and immuno-fluorescence staining. RESULTS Pretreating the mice with CRex once at 1,000 or 3,000 mg/kg and twice at 1,000 mg/kg 1 h before MCAO, brought about a significantly decrease in the infarction volumes. Furthermore, pretreating mice with CRex once at 3,000 mg/kg 1 h before MCAO significantly suppressed the reduction of forepaw grip strength of MCAO-induced mice. In the MCAO-induced group, preadministration of CRex inhibited the reduction in the discrimination ratio brought on by MCAO in a similar manner. CRex exhibited these effects by suppressing the activation of astrocytes and microglia, which regulated the inflammatory response. CONCLUSIONS This study proposes a novel development for the treatment of ischemic stroke and provides evidence favoring the use of L. chuanxiong rhizomes against ischemic stroke.
Collapse
Affiliation(s)
- Chiyeon Lim
- College of Medicine, Dongguk University, Goyang, 10326, Republic of Korea
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Sehyun Lim
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
- School of Public Health, Far East University, Eumseong, 27601, Republic of Korea
| | - So-Jung Moon
- College of Science & Industry Convergence, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Suin Cho
- School of Korean Medicine, Pusan National University, Yangsan, 50612, Republic of Korea.
- Department of Korean Medicine, School of Korean Medicine, Yangsan Campus of Pusan National University, Yangsan, 50612, Republic of Korea.
| |
Collapse
|
4
|
Chen X, Chen A, Wei J, Huang Y, Deng J, Chen P, Yan Y, Lin M, Chen L, Zhang J, Huang Z, Zeng X, Gong C, Zheng X. Dexmedetomidine alleviates cognitive impairment by promoting hippocampal neurogenesis via BDNF/TrkB/CREB signaling pathway in hypoxic-ischemic neonatal rats. CNS Neurosci Ther 2024; 30:e14486. [PMID: 37830170 PMCID: PMC10805444 DOI: 10.1111/cns.14486] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 09/19/2023] [Accepted: 09/20/2023] [Indexed: 10/14/2023] Open
Abstract
AIMS Dexmedetomidine (DEX) has been reported to alleviate hypoxic-ischemic brain damage (HIBD) in neonates. This study aimed to investigate whether DEX improves cognitive impairment by promoting hippocampal neurogenesis via the BDNF/TrkB/CREB signaling pathway in neonatal rats with HIBD. METHODS HIBD was induced in postnatal day 7 rats using the Rice-Vannucci method, and DEX (25 μg/kg) was administered intraperitoneally immediately after the HIBD induction. The BDNF/TrkB/CREB pathway was regulated by administering the TrkB receptor antagonist ANA-12 through intraperitoneal injection or by delivering adeno-associated virus (AAV)-shRNA-BDNF via intrahippocampal injection. Western blot was performed to measure the levels of BDNF, TrkB, and CREB. Immunofluorescence staining was utilized to identify the polarization of astrocytes and evaluate the levels of neurogenesis in the dentate gyrus of the hippocampus. Nissl and TTC staining were performed to evaluate the extent of neuronal damage. The MWM test was conducted to evaluate spatial learning and memory ability. RESULTS The levels of BDNF and neurogenesis exhibited a notable decrease in the hippocampus of neonatal rats after HIBD, as determined by RNA-sequencing technology. Our results demonstrated that treatment with DEX effectively increased the protein expression of BDNF and the phosphorylation of TrkB and CREB, promoting neurogenesis in the dentate gyrus of the hippocampus in neonatal rats with HIBD. Specifically, DEX treatment significantly augmented the expression of BDNF in hippocampal astrocytes, while decreasing the proportion of detrimental A1 astrocytes and increasing the proportion of beneficial A2 astrocytes in neonatal rats with HIBD. Furthermore, inhibiting the BDNF/TrkB/CREB pathway using either ANA-12 or AAV-shRNA-BDNF significantly counteracted the advantageous outcomes of DEX on hippocampal neurogenesis, neuronal survival, and cognitive improvement. CONCLUSIONS DEX promoted neurogenesis in the hippocampus by activating the BDNF/TrkB/CREB pathway through the induction of polarization of A1 astrocytes toward A2 astrocytes, subsequently mitigating neuronal damage and cognitive impairment in neonates with HIBD.
Collapse
Affiliation(s)
- Xiaohui Chen
- Department of AnesthesiologyShengli Clinical Medical College of Fujian Medical University, Fujian Provincial HospitalFuzhouChina
| | - Andi Chen
- Department of AnesthesiologyShengli Clinical Medical College of Fujian Medical University, Fujian Provincial HospitalFuzhouChina
| | - Jianjie Wei
- Department of AnesthesiologyShengli Clinical Medical College of Fujian Medical University, Fujian Provincial HospitalFuzhouChina
| | - Yongxin Huang
- Department of AnesthesiologyShengli Clinical Medical College of Fujian Medical University, Fujian Provincial HospitalFuzhouChina
| | - Jianhui Deng
- Department of AnesthesiologyShengli Clinical Medical College of Fujian Medical University, Fujian Provincial HospitalFuzhouChina
| | - Pinzhong Chen
- Department of AnesthesiologyShengli Clinical Medical College of Fujian Medical University, Fujian Provincial HospitalFuzhouChina
| | - Yanlin Yan
- Department of AnesthesiologyShengli Clinical Medical College of Fujian Medical University, Fujian Provincial HospitalFuzhouChina
| | - Mingxue Lin
- Department of AnesthesiologyShengli Clinical Medical College of Fujian Medical University, Fujian Provincial HospitalFuzhouChina
| | - Lifei Chen
- Department of AnesthesiologyShengli Clinical Medical College of Fujian Medical University, Fujian Provincial HospitalFuzhouChina
| | - Jiuyun Zhang
- Fujian Provincial Key Laboratory of Emergency MedicineFuzhouChina
| | - Zhibin Huang
- Department of AnesthesiologyShengli Clinical Medical College of Fujian Medical University, Fujian Provincial HospitalFuzhouChina
| | - Xiaoqian Zeng
- Department of AnesthesiologyShengli Clinical Medical College of Fujian Medical University, Fujian Provincial HospitalFuzhouChina
| | - Cansheng Gong
- Department of AnesthesiologyShengli Clinical Medical College of Fujian Medical University, Fujian Provincial HospitalFuzhouChina
| | - Xiaochun Zheng
- Department of AnesthesiologyShengli Clinical Medical College of Fujian Medical University, Fujian Provincial HospitalFuzhouChina
- Fujian Provincial Key Laboratory of Emergency MedicineFuzhouChina
- Fujian Emergency Medical Center, Fujian Provincial Key Laboratory of Critical Care MedicineFujian Provincial Co‐Constructed Laboratory of “Belt and Road”FuzhouChina
| |
Collapse
|
5
|
Tan X, Zhang K, Shi W, Tang Z. Research progress on the regulation and mechanism of borneol on the blood-brain barrier in pathological states: a narrative review focused on ischemic stroke and cerebral glioma. Transl Cancer Res 2023; 12:3198-3209. [PMID: 38130309 PMCID: PMC10731338 DOI: 10.21037/tcr-23-1487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 10/12/2023] [Indexed: 12/23/2023]
Abstract
Background and Objective The blood-brain barrier (BBB) serves as a dynamic, selective shield, safeguarding the central nervous system (CNS) by separating the brain from circulating blood, preserving its microenvironment, and ensuring stability. However, in the presence of brain pathology, drug delivery across the BBB and blood-tumor barrier (BTB) becomes challenging, hindering effective treatments. Borneol exhibits promise in bidirectionally modulating the BBB under pathological conditions, suggesting at potential clinical applications for related diseases. Our primary goal in this review is to investigate borneol's potential clinical utility in bidirectionally regulating the BBB under pathological conditions. Methods The PubMed database, CNKI (China National Knowledge Infrastructure), Wanfang Data were searched to retrieve articles on animal experiments and cell-based research published from January 1, 2003, to May 1, 2023, using the following medical subject headings (MeSH) terms: borneol, blood-brain barrier, ischemic stroke, cerebral gliomas, anti-inflammatory. The search was limited to articles published in English and Chinese. In total, 86 articles were deemed eligible for inclusion in this study. Key Content and Findings The breakdown of the BBB is a key pathological process in ischemic stroke and cerebral glioma. When used alone, the lipophilic properties of borneol can reduce the permeability of the BBB and restore its normal function, thereby repairing brain damage and protecting brain tissue. Its specific protective effects may be related to inflammatory regulation mechanisms. The anti-inflammatory and protective effects of borneol can be used to improve and treat lesions caused by ischemic stroke and cerebral glioma. Furthermore, when combined with other drugs, borneol can accelerate the opening of the BBB, improve permeability through physiological processes, and enhance drug penetration and distribution in the brain without causing pathological damage to the brain. Conclusions This review summarizes the mechanisms by which borneol regulates the BBB and BTB in ischemic stroke and cerebral glioma, and discusses the potential clinical applications of borneol in the treatment of these diseases. It is believed that in the future, as research methods are refined, more effective and targeted therapies for cerebral glioma and ischemic stroke will be explored related to the protective mechanism of the BBB under pathological conditions with borneol alone or in combination with other drugs.
Collapse
Affiliation(s)
- Xinghua Tan
- Department of Pharmacy, Shaoxing People’s Hospital, Shaoxing, China
| | - Ke Zhang
- Department of Pharmacy, Shaoxing People’s Hospital, Shaoxing, China
| | - Wenyin Shi
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Zhihua Tang
- Department of Pharmacy, Shaoxing People’s Hospital, Shaoxing, China
| |
Collapse
|
6
|
Long J, Sun Y, Liu S, Yang S, Chen C, Zhang Z, Chu S, Yang Y, Pei G, Lin M, Yan Q, Yao J, Lin Y, Yi F, Meng L, Tan Y, Ai Q, Chen N. Targeting pyroptosis as a preventive and therapeutic approach for stroke. Cell Death Discov 2023; 9:155. [PMID: 37165005 PMCID: PMC10172388 DOI: 10.1038/s41420-023-01440-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 04/18/2023] [Accepted: 04/20/2023] [Indexed: 05/12/2023] Open
Abstract
Stroke has caused tremendous social stress worldwide, yet despite decades of research and development of new stroke drugs, most have failed and rt-PA (Recombinant tissue plasminogen activator) is still the accepted treatment for ischemic stroke. the complexity of the stroke mechanism has led to unsatisfactory efficacy of most drugs in clinical trials, indicating that there are still many gaps in our understanding of stroke. Pyroptosis is a programmed cell death (PCD) with inflammatory properties and are thought to be closely associated with stroke. Pyroptosis is regulated by the GSDMD of the gasdermin family, which when cleaved by Caspase-1/Caspase-11 into N-GSDMD with pore-forming activity can bind to the plasma membrane to form small 10-20 nm pores, which would allow the release of inflammatory factors IL-18 and IL-1β before cell rupture, greatly exacerbating the inflammatory response. The pyroptosis occurs mainly in the border zone of cerebral infarction, and glial cells, neuronal cells and brain microvascular endothelial cells (BMECs) all undergo pyroptosis after stroke, which largely exacerbates the breakdown of the blood-brain barrier (BBB) and thus aggravates brain injury. Therefore, pyroptosis may be a good direction for the treatment of stroke. In this review, we focus on the latest mechanisms of action of pyroptosis and the process by which pyroptosis regulates stroke development. We also suggest potential therapeutic stroke drugs that target the pyroptosis pathway, providing additional therapeutic strategies for the clinical management of stroke. The role of pyroptosis after stroke. After stroke, microglia first rush to the damaged area and polarize into M1 and M2 types. Under the influence of various stimuli, microglia undergo pyroptosis, release pro-inflammatory factors, and are converted to the M1 type; astrocytes and neuronal cells also undergo pyroptosis under the stimulation of various pro-inflammatory factors, leading to astrocyte death due to increased osmotic pressure in the membrane, resulting in water absorption and swelling until rupture. BMECs, the main structural component of the BBB, also undergo pyroptosis when stimulated by pro-inflammatory factors released from microglia and astrocytes, leading to the destruction of the structural integrity of the BBB, ultimately causing more severe brain damage. In addition, GSDMD in neutrophils mainly mediate the release of NETs rather than pyroptosis, which also aggravates brain injury. IL-10=interleukin-10; TGF-β = transforming growth factor-β; IL-18=interleukin-18; IL-1β = interleukin-1β; TNF-α = tumor necrosis factor-α; iNOS=induced nitrogen monoxide synthase; MMPs=Matrix metalloproteinases; GSDMD = gasdermin D; BMECs=brain microvascular endothelial cells; BBB = blood-brain barrier.
Collapse
Affiliation(s)
- Junpeng Long
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha, P. R. China
| | - Yang Sun
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha, P. R. China
| | - Shasha Liu
- Department of Pharmacy, Changsha Hospital for Matemal & Child Health Care, Changsha, P. R. China
| | - Songwei Yang
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha, P. R. China
| | - Chen Chen
- Department of Pharmacy, The First Hospital of Lanzhou University, Lanzhou, P. R. China
| | - Zhao Zhang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Shifeng Chu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Yantao Yang
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha, P. R. China
| | - Gang Pei
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha, P. R. China
| | - Meiyu Lin
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha, P. R. China
| | - Qian Yan
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha, P. R. China
| | - Jiao Yao
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha, P. R. China
| | - Yuting Lin
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha, P. R. China
| | - Fan Yi
- Key Laboratory of Cosmetic, China National Light Industry, Beijing Technology and Business University, Beijing, P. R. China
| | - Lei Meng
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha, P. R. China
| | - Yong Tan
- Department of Nephrology, Xiangtan Central Hospital, Xiangtan, P. R. China
| | - Qidi Ai
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha, P. R. China.
| | - Naihong Chen
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha, P. R. China.
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China.
| |
Collapse
|
7
|
Gao F, Chen R, Li S, Li A, Bai B, Mi R, Xue G. (+)-Borneol exerts neuroprotective effects via suppressing the NF-κB pathway in the pilocarpine-induced epileptogenesis rat model. Brain Res 2023; 1810:148382. [PMID: 37127175 DOI: 10.1016/j.brainres.2023.148382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 04/25/2023] [Accepted: 04/28/2023] [Indexed: 05/03/2023]
Abstract
Neuroinflammation plays a crucial role in the development of epilepsy, and suppressing neuroinflammation can delay epileptogenesis. Recent reports have demonstrated that (+)-borneol has neuroprotective effects in several brain disorders by reducing neuroinflammation. However, its effects on epilepsy have not been reported. In this research, we first studied the effect of different doses of (+)-borneol (3, 6, and 12 mg/kg) on neuroinflammation in a pilocarpine model of epileptogenesis by detecting IL-1β, TNF-α, and COX-2 expression. We demonstrated that different doses of (+)-borneol decreased IL-1β, TNF-α, and COX-2 levels, with 12 mg/kg having the most substantial effect. Furthermore, we examined the effects of 12 mg/kg (+)-borneol on neuronal damage, glial cell activation, and apoptosis in the hippocampus at different time points (1, 3, and 7 days) after SE. We found that (+)-borneol significantly ameliorated neuronal injury, decreased glial cell activation, and attenuated apoptosis. We also found that (+)-borneol inhibited the NF-κB pathway activation induced by SE. In conclusion, our results indicated that (+)-borneol reduces neuroinflammation by inhibiting the NF-κB pathway activation, exerts neuroprotective effects, and may have an inhibitory effect in epileptogenesis.
Collapse
Affiliation(s)
- Fankai Gao
- Department of Neurology, The Second Hospital of Shanxi Medical University, No. 382 Wuyi Road, Taiyuan, 030001, Shanxi Province, China
| | - Rui Chen
- Department of Neurology, The Second Hospital of Shanxi Medical University, No. 382 Wuyi Road, Taiyuan, 030001, Shanxi Province, China
| | - Shuo Li
- Department of Neurology, The Second Hospital of Shanxi Medical University, No. 382 Wuyi Road, Taiyuan, 030001, Shanxi Province, China
| | - An Li
- Department of Neurology, The Second Hospital of Shanxi Medical University, No. 382 Wuyi Road, Taiyuan, 030001, Shanxi Province, China
| | - Bo Bai
- Department of Neurology, The Second Hospital of Shanxi Medical University, No. 382 Wuyi Road, Taiyuan, 030001, Shanxi Province, China
| | - Rulin Mi
- Department of Neurology, The Second Hospital of Shanxi Medical University, No. 382 Wuyi Road, Taiyuan, 030001, Shanxi Province, China
| | - Guofang Xue
- Department of Neurology, The Second Hospital of Shanxi Medical University, No. 382 Wuyi Road, Taiyuan, 030001, Shanxi Province, China.
| |
Collapse
|
8
|
Łuszczki JJ, Bojar H, Góralczyk A, Skalicka-Woźniak K. Antiseizure Effects of Scoparone, Borneol and Their Impact on the Anticonvulsant Potency of Four Classic Antiseizure Medications in the Mouse MES Model-An Isobolographic Transformation. Int J Mol Sci 2023; 24:ijms24021395. [PMID: 36674911 PMCID: PMC9867083 DOI: 10.3390/ijms24021395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 01/05/2023] [Accepted: 01/06/2023] [Indexed: 01/13/2023] Open
Abstract
Numerous botanical drugs containing coumarins and terpenes are used in ethnomedicine all over the world for their various therapeutic properties, especially those affecting the CNS system. The treatment of epilepsy is based on antiseizure medications (ASMs), although novel strategies using naturally occurring substances with confirmed antiseizure properties are being developed nowadays. The aim of this study was to determine the anticonvulsant profiles of scoparone (a simple coumarin) and borneol (a bicyclic monoterpenoid) when administered separately and in combination, as well as their impact on the antiseizure effects of four classic ASMs (carbamazepine, phenytoin, phenobarbital and valproate) in the mouse model of maximal electroshock-induced (MES) tonic-clonic seizures. MES-induced seizures were evoked in mice receiving the respective doses of the tested natural compounds and classic ASMs (when applied alone or in combinations). Interactions for two-drug and three-drug mixtures were assessed by means of isobolographic transformation of data. Polygonograms were used to illustrate the types of interactions occurring among drugs. The total brain content of ASMs was measured in mice receiving the respective drug treatments with fluorescent polarization immunoassay. Scoparone and borneol, when administered alone, exerted anticonvulsant properties in the mouse MES model. The two-drug mixtures of scoparone with valproate, borneol with phenobarbital and borneol with valproate produced synergistic interactions in the mouse MES model, while the remaining tested two-drug mixtures produced additivity. The three-drug mixtures of scoparone + borneol with valproate and phenobarbital produced synergistic interactions in the mouse MES model. Verification of total brain concentrations of valproate and phenobarbital revealed that borneol elevated the total brain concentrations of both ASMs, while scoparone did not affect the brain content of these ASMs in mice. The synergistic interaction of scoparone with valproate observed in the mouse MES model is pharmacodynamic in nature. Borneol elevated the brain concentrations of the tested ASMs, contributing to the pharmacokinetic nature of the observed synergistic interactions with valproate and phenobarbital in the mouse MES model.
Collapse
Affiliation(s)
- Jarogniew J. Łuszczki
- Department of Occupational Medicine, Medical University of Lublin, 20-090 Lublin, Poland
- Correspondence: ; Tel.: +48-81-448-6500; Fax: +48-81-448-6501
| | - Hubert Bojar
- Department of Toxicology and Food Safety, Institute of Rural Health, 20-090 Lublin, Poland
| | - Agnieszka Góralczyk
- Department of Occupational Medicine, Medical University of Lublin, 20-090 Lublin, Poland
| | | |
Collapse
|
9
|
Zhang Y, Liu Q, Zhang T, Wang H, Fu Y, Wang W, Li D. The therapeutic role of Jingchuan tablet on ischaemic cerebral stroke via the HIF-1α/EPO/VEGFA signalling pathway. PHARMACEUTICAL BIOLOGY 2022; 60:2110-2123. [PMID: 36269045 PMCID: PMC9590438 DOI: 10.1080/13880209.2022.2134430] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 06/20/2022] [Accepted: 09/30/2022] [Indexed: 06/03/2023]
Abstract
CONTEXT Jingchuan tablet (JCT) is a Chinese medicine prescription for treating ischaemic cerebral stroke (ICS). However, its relevant mechanisms remain unclear. OBJECTIVE To unravel the intrinsic mechanisms of JCT anti-ICS. MATERIALS AND METHODS 'Hongjingtian', 'chuanxiong', 'yanhusuo', 'bingpian', 'cerebral infarction', 'cerebral ischemia' or 'stroke' were used as keywords, and then components, targets and underlying mechanisms of JCT anti-ICS were analysed in TCMSP, TTD, DrugBank, STRING and Metascape databases up to June 2020. Male Sprague-Dawley rats under permanent middle cerebral artery occlusion (pMCAO) model, randomly assigned as: model, sham, nimodipine (0.012 g/kg/d) and JCT (0.78, 1.56 and 3.12 g/kg/d) groups, received oral gavage administration for a week. Therapeutic effects were evaluated by detecting the proportion of cerebral infarction, neuronal apoptosis and neurological deficits. Bioactive components were detected by HPLC-MS. Molecular biology and computational docking were used to verify the underlying mechanisms. RESULTS Eighty-one components, 166 targets and HIF-1α/EPO/VEGFA pathway contributed to the anti-ICS effect of JCT. JCT treatment effectively reduced the proportion of cerebral infarction (33.13%), apoptosis rate (14.80%) and neurobehavioural score (2.00). JCT increased the protein levels of HIF-1α (0.84), EPO (0.64) and VEGFA (0.69), respectively (p < 0.05). Gallic acid, salidroside, chlorogenic acid, ethyl gallate, ferulic acid and tetrahydropalmatine detected by HPLC-MS showed good interaction and binding with HIF-1α/EPO/VEGFA. CONCLUSIONS Our study demonstrated the mechanisms of JCT anti-ICS associated with the activation of the HIF-1α/EPO/VEGFA pathway, which provided a pharmacological basis for expanding the clinical application and some scientific ideas for further research into the material basis JCT anti-ICS.
Collapse
Affiliation(s)
- Yan Zhang
- Tianjin Institute of Medical and Pharmaceutical Sciences, Tianjin, China
| | - Qinghuan Liu
- Tianjin Institute of Medical and Pharmaceutical Sciences, Tianjin, China
| | - Ting Zhang
- Tianjin Institute of Medical and Pharmaceutical Sciences, Tianjin, China
| | - Hong Wang
- Tianjin Institute of Medical and Pharmaceutical Sciences, Tianjin, China
| | - Yu Fu
- Tianjin Institute of Medical and Pharmaceutical Sciences, Tianjin, China
| | - Wentong Wang
- Tianjin Institute of Medical and Pharmaceutical Sciences, Tianjin, China
| | - Dongdong Li
- Tianjin Institute of Medical and Pharmaceutical Sciences, Tianjin, China
| |
Collapse
|
10
|
Sun L, Ye X, Wang L, Yu J, Wu Y, Wang M, Dai L. A Review of Traditional Chinese Medicine, Buyang Huanwu Decoction for the Treatment of Cerebral Small Vessel Disease. Front Neurosci 2022; 16:942188. [PMID: 35844225 PMCID: PMC9278698 DOI: 10.3389/fnins.2022.942188] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 06/02/2022] [Indexed: 11/13/2022] Open
Abstract
Cerebral small vessel disease (CSVD) is often referred to as “collaterals disease” in traditional Chinese medicine (TCM), and commonly includes ischemic and hemorrhagic CSVD. TCM has a long history of treating CSVD and has demonstrated unique efficacy. Buyang Huanwu Decoction (BHD) is a classical TCM formula that has been used for the prevention and treatment of stroke for hundreds of years. BHD exerts its therapeutic effects on CSVD through a variety of mechanisms. In this review, the clinical and animal studies on BHD and CSVD were systematically introduced. In addition, the pharmacological mechanisms, active components, and clinical applications of BHD in the treatment of CSVD were reviewed. We believe that an in-depth understanding of BHD, its pharmacological mechanism, disease-drug interaction, and other aspects will help in laying the foundation for its development as a new therapeutic strategy for the treatment of CSVD.
Collapse
|